1
|
Wen Y, Sparks Z, Hawkins I, Lednicky J, Abboud G, Nelson C, Chauhan A, Driver J. Sustained release system from PLGA particles co-encapsulated with inactivated influenza virus with natural killer T cell agonist α-galactosylceramide. Eur J Pharm Biopharm 2024; 201:114365. [PMID: 38876362 DOI: 10.1016/j.ejpb.2024.114365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/18/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Vaccines against influenza and many other infectious diseases require multiple boosters in addition to the primary dose to improve efficacy, but this approach is not ideal for compliance. The multiple doses could potentially be replaced by sustained or pulsatile release of antigens encapsulated in degradable microparticles (MPs). The efficacy of a vaccine is improved by adding an adjuvant, which can be co-delivered from the particles to enhance immunogenicity. Here, we developed degradable poly-lactic-co-glycolic acid (PLGA) (7-17 kDa) MPs capable of sustained release of ultraviolet killed influenza virus (A/PR/8/34) (kPR8) vaccine and the natural killer T (NKT) cell agonist alpha-galactosylceramide (α-GalCer) and tested their effectiveness at providing long-term protection against influenza virus infection in mice. Multiple formulations were developed for encapsulating the virus and adjuvant separately, and in combination. The MPs exhibited sustained release of both the virus and the adjuvant lasting more than a month. Co-encapsulation significantly increased the encapsulation efficiency (EE) of the vaccine but reduced the release duration. On the other hand, co-encapsulation led to a reduction in EE for the α-GalCer and a change in release profile to a higher initial burst followed by a linear release compared to a low initial burst and slower linear release. The α-GalCer also had considerably longer release duration compared to the vaccine. Mice injected with particle formulations co-encapsulating kPR8 and α-GalCer were protected from a lethal influenza virus infection 30 weeks after vaccination. This study demonstrates that PLGA MP based vaccines are promising for providing effective vaccination and possibly for replacing multiple doses with a single injection.
Collapse
Affiliation(s)
- Yuhan Wen
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, United States
| | - Zachary Sparks
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO 80401, United States
| | - Ian Hawkins
- Department of Comparative, Diagnostic & Population Medicine, University of Florida, Gainesville, FL 32612, United States
| | - John Lednicky
- Department of Environmental and Global Health, University of Florida, Gainesville, FL 32612, United States
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32612, United States
| | - Corwin Nelson
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, United States
| | - Anuj Chauhan
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO 80401, United States.
| | - John Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO 65201, United States; Bond Life Sciences Center, University of Missouri, Columbia, MO 65201, United States.
| |
Collapse
|
2
|
Hasija M, Ma J, Li B, Rahman N, Strahlendorf KA, Ausar SF. Determination of Deamidation in Adjuvanted Vaccine Antigens through Isoaspartic Acid Quantification. Vaccines (Basel) 2024; 12:733. [PMID: 39066371 PMCID: PMC11281650 DOI: 10.3390/vaccines12070733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Deamidation is a post-translational chemical modification that occurs within proteins and can be influenced by many factors, including temperature and pH. In vaccines, deamidation is considered undesirable as it may lead to changes in structure, function, stability, and immunogenicity. Detecting deamidation in vaccines, especially adjuvanted vaccines, can be challenging due to the lack of simple quantitative techniques. In this study, the quantification of isoaspartic acid (isoAsp) was used to assess deamidation in model antigens in the presence and absence of common vaccine adjuvants. This study shows that the detection of isoAsp was possible in the presence of various types of adjuvants with little to no interference. High levels of isoAsp were detected in thermally and pH-stressed adjuvanted vaccines, suggesting significant deamidation and highlighting the stability-indicating capabilities of the assay. The quantification of isoAsp in stability programs of a vaccine drug product could possibly find applications in product shelf-life determination, using thermal kinetic modeling to predict deamidation over time. The ability to detect deamidation early in vaccine development enhances process improvements and ultimately improves the vaccine's stability. To summarize, this paper describes a rapid and simple method to determine deamidation in adjuvanted vaccines. This method could be applicable to formulation development, stability assessment, or shelf-life determination.
Collapse
|
3
|
Lavelle EC, McEntee CP. Vaccine adjuvants: Tailoring innate recognition to send the right message. Immunity 2024; 57:772-789. [PMID: 38599170 DOI: 10.1016/j.immuni.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Adjuvants play pivotal roles in vaccine development, enhancing immunization efficacy through prolonged retention and sustained release of antigen, lymph node targeting, and regulation of dendritic cell activation. Adjuvant-induced activation of innate immunity is achieved via diverse mechanisms: for example, adjuvants can serve as direct ligands for pathogen recognition receptors or as inducers of cell stress and death, leading to the release of immunostimulatory-damage-associated molecular patterns. Adjuvant systems increasingly stimulate multiple innate pathways to induce greater potency. Increased understanding of the principles dictating adjuvant-induced innate immunity will subsequently lead to programming specific types of adaptive immune responses. This tailored optimization is fundamental to next-generation vaccines capable of inducing robust and sustained adaptive immune memory across different cohorts.
Collapse
Affiliation(s)
- Ed C Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Craig P McEntee
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Ribette T, Charretier Y, Laurent S, Syntin P, Chautard E, Meniche X, Darnaud M, Bequet F, Beloeil L, Piras-Douce F, Abi-Ghanem J. Development of Mass Spectrometry Imaging on skeletal muscle to characterize the local pro-inflammatory and pro-resolution lipid responses in a vaccination context. J Proteomics 2024; 296:105105. [PMID: 38325731 DOI: 10.1016/j.jprot.2024.105105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
Vaccine reactogenicity is well documented at the clinical level but the mechanism involved at the local or systemic level are still poorly understood. Muscular tissue where most vaccines are administered is the first place of interaction between the vaccine formulation and the host's immune cells. So far, this site of vaccine administration is not well documented from a mechanistic standpoint. The study of early molecular events at the injection site is crucial to understand the local response to vaccines. In this paper, we report a standardized workflow, from the injection of vaccine formulations in rabbit muscle, to the analysis by desorption electrospray ionization and histology staining to understand the role of lipids involved in the inflammation and its resolution on striated muscular tissue. The analysis of lipid mediators was optimized at the site of needle insertion to allow the spatial comparison of cellular infiltrates at the injection site. We showed that lipids were distributed across the spatial tissue morphology in a time-dependent manner. The MS imaging applied to vaccinology could pave the way to a better understanding of vaccine reactogenicity and mechanism of action.
Collapse
|
5
|
Huang Z, Gong H, Sun Q, Yang J, Yan X, Xu F. Research progress on emulsion vaccine adjuvants. Heliyon 2024; 10:e24662. [PMID: 38317888 PMCID: PMC10839794 DOI: 10.1016/j.heliyon.2024.e24662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Vaccination is the most cost-effective method for preventing various infectious diseases. Compared with conventional vaccines, new-generation vaccines, especially recombinant protein or synthetic peptide vaccines, are safer but less immunogenic than crude inactivated microbial vaccines. The immunogenicity of these vaccines can be enhanced using suitable adjuvants. This is the main reason why adjuvants are of great importance in vaccine development. Several novel human emulsion-based vaccine adjuvants (MF59, AS03) have been approved for clinical use. This paper reviews the research progress on emulsion-based adjuvants and focuses on their mechanism of action. An outlook can be provided for the development of emulsion-based vaccine adjuvants.
Collapse
Affiliation(s)
- Zhuanqing Huang
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Hui Gong
- Medical School of Chinese PLA, Beijing 100853, China
| | - Qi Sun
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Jinjin Yang
- The Fifth medical center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xiaochuan Yan
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
| | - Fenghua Xu
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| |
Collapse
|
6
|
Shichinohe S, Watanabe T. Advances in Adjuvanted Influenza Vaccines. Vaccines (Basel) 2023; 11:1391. [PMID: 37631959 PMCID: PMC10459454 DOI: 10.3390/vaccines11081391] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/29/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023] Open
Abstract
The numerous influenza infections that occur every year present a major public health problem. Influenza vaccines are important for the prevention of the disease; however, their effectiveness against infection can be suboptimal. Particularly in the elderly, immune induction can be insufficient, and the vaccine efficacy against infection is usually lower than that in young adults. Vaccine efficacy can be improved by the addition of adjuvants, and an influenza vaccine with an oil-in-water adjuvant MF59, FLUAD, has been recently licensed in the United States and other countries for persons aged 65 years and older. Although the adverse effects of adjuvanted vaccines have been a concern, many adverse effects of currently approved adjuvanted influenza vaccines are mild and acceptable, given the overriding benefits of the vaccine. Since sufficient immunity can be induced with a small amount of vaccine antigen in the presence of an adjuvant, adjuvanted vaccines promote dose sparing and the prompt preparation of vaccines for pandemic influenza. Adjuvants not only enhance the immune response to antigens but can also be effective against antigenically different viruses. In this narrative review, we provide an overview of influenza vaccines, both past and present, before presenting a discussion of adjuvanted influenza vaccines and their future.
Collapse
Grants
- JP16H06429, JP16K21723, JP17H05809, JP16H06434, JP22H02521, JP22H02876 Japan Society for the Promotion of Science
- JP20jk0210021h0002, JP19fk0108113, JP223fa627002, JP22am0401030, JP23fk0108659, JP22gm1610010 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Shintaro Shichinohe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tokiko Watanabe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease and Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
7
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
8
|
Filipić B, Pantelić I, Nikolić I, Majhen D, Stojić-Vukanić Z, Savić S, Krajišnik D. Nanoparticle-Based Adjuvants and Delivery Systems for Modern Vaccines. Vaccines (Basel) 2023; 11:1172. [PMID: 37514991 PMCID: PMC10385383 DOI: 10.3390/vaccines11071172] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Ever since the development of the first vaccine, vaccination has had the great impact on global health, leading to the decrease in the burden of numerous infectious diseases. However, there is a constant need to improve existing vaccines and develop new vaccination strategies and vaccine platforms that induce a broader immune response compared to traditional vaccines. Modern vaccines tend to rely on certain nanotechnology platforms but are still expected to be readily available and easy for large-scale manufacturing and to induce a durable immune response. In this review, we present an overview of the most promising nanoadjuvants and nanoparticulate delivery systems and discuss their benefits from tehchnological and immunological standpoints as well as their objective drawbacks and possible side effects. The presented nano alums, silica and clay nanoparticles, nanoemulsions, adenoviral-vectored systems, adeno-associated viral vectors, vesicular stomatitis viral vectors, lentiviral vectors, virus-like particles (including bacteriophage-based ones) and virosomes indicate that vaccine developers can now choose different adjuvants and/or delivery systems as per the requirement, specific to combatting different infectious diseases.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
- Section of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| |
Collapse
|
9
|
Ustyugova IV, Pougatcheva S, Farrell T, Strugnell T, Ganesh V, Zeldovich KB, Chivukula S, Goncalvez AP, Barro M. AF03 adjuvant improves anti-hemagglutinin and anti-neuraminidase immune responses induced by licensed seasonal quadrivalent influenza vaccines in mice. Vaccine 2023; 41:2022-2034. [PMID: 36803901 DOI: 10.1016/j.vaccine.2023.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/21/2023]
Abstract
Seasonal influenza remains a serious public health concern as the viral infection spreads easily from person to person and due to antigenic drift of neutralizing epitopes. Vaccination is the best method for disease prevention, however current seasonal influenza vaccines stimulate antibodies which are often effective against only antigenically similar strains. To boost the immune responses and increase vaccine effectiveness, adjuvants have been used for the past 20 years. The current study explores the use of oil-in-water adjuvant, AF03 to improve an immunogenicity of 2 licensed vaccines. A standard-dose inactivated quadrivalent influenza vaccine (IIV4-SD), containing both hemagglutinin (HA) and neuraminidase (NA) antigens, and recombinant quadrivalent influenza vaccine (RIV4), containing only HA-antigen were adjuvanted with AF03 in naïve BALB/c mouse model. Functional HA-specific antibody titers against all four homologous vaccine strains were enhanced by AF03, indicating potential increase in protective immunity. An increase in HA-specific total immunoglobulin G (IgG) binding titers were detected against homologous HAs, heterologous panel of 30 H3 HAs and seven Influenza B HAs. The neuraminidase inhibition (NAI) activity was significantly higher in IIV4-SD-AF03 group. Use of AF03 adjuvant improved the immune response to two influenza vaccines in a mouse model via an increase in functional and total antibodies against NA and a broad panel of HA-antigens.
Collapse
|
10
|
Syntin P, Piras-Douce F, Dalençon F, Garinot M, Haensler J. Nonclinical safety assessments of a novel synthetic toll-like receptor 4 agonist and saponin based adjuvant. Toxicol Appl Pharmacol 2023; 460:116358. [PMID: 36572229 DOI: 10.1016/j.taap.2022.116358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
A full nonclinical safety package was performed to support the clinical use of SPA14, a novel liposome-based vaccine adjuvant containing the synthetic toll-like receptor 4 agonist E6020 and saponin QS21. E6020 and QS21 were tested negative for their potential genotoxic effects in Ames, micronucleus, or mouse-lymphoma TK (thymidine kinase) assay. To evaluate the potential local and systemic effects of SPA14, two toxicity studies were performed in rabbits. In the first dose range finding toxicity study, rabbits received two intramuscular injections of SPA14 at increasing doses of E6020 combined with two antigens, a control (saline), the two antigens alone, or the antigens adjuvanted with a liposome-based adjuvant AS01B. No systemic toxicity was detected, supporting the dose of 5 μg of E6020 for the subsequent pivotal study. In the second repeated dose toxicity study, rabbits received four intramuscular injections of SPA14 alone, a control (saline), SPA14 combined with two antigens, the two antigens alone, or the antigens combined with AF03 adjuvant, which is a squalene-based emulsion. SPA14 alone or in combination with the antigens was well tolerated and did not cause any systemic toxicity. Finally, two safety pharmacology studies were conducted to assess potential cardiovascular and respiratory effects of E6020 and SPA14 in conscious telemetered cynomolgus monkeys and beagle dogs, respectively. One subcutaneous injection of E6020 in monkeys and one intramuscular injection of SPA14 in dogs had no consequences on respiratory and cardiovascular functions. Altogether these results support the clinical development of SPA14.
Collapse
|
11
|
Zeng Y, Zou F, Xia N, Li S. In-depth review of delivery carriers associated with vaccine adjuvants: current status and future perspectives. Expert Rev Vaccines 2023; 22:681-695. [PMID: 37496496 DOI: 10.1080/14760584.2023.2238807] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Vaccines are powerful tools for controlling microbial infections and preventing epidemics. To enhance the immune response to antigens, effective subunit vaccines or mRNA vaccines often require the combination of adjuvants or delivery carriers. In recent years, with the rapid development of immune mechanism research and nanotechnology, various studies based on the optimization of traditional adjuvants or various novel carriers have been intensified, and the construction of vaccine adjuvant delivery systems (VADS) with both adjuvant activity and antigen delivery has become more and more important in vaccine research. AREAS COVERED This paper reviews the common types of vaccine adjuvant delivery carriers, classifies the VADS according to their basic carrier types, introduces the current research status and future development trend, and emphasizes the important role of VADS in novel vaccine research. EXPERT OPINION As the number of vaccine types increases, conventional aluminum adjuvants show limitations in effectively stimulating cellular immune responses, limiting their use in therapeutic vaccines for intracellular infections or tumors. In contrast, the use of conventional adjuvants as VADS to carry immunostimulatory molecules or deliver antigens can greatly enhance the immune boosting effect of classical adjuvants. A comprehensive understanding of the various delivery vehicles will further facilitate the development of vaccine adjuvant research.
Collapse
Affiliation(s)
- Yarong Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Feihong Zou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
Immunogenicity and protective efficacy of RSV G central conserved domain vaccine with a prefusion nanoparticle. NPJ Vaccines 2022; 7:74. [PMID: 35773301 PMCID: PMC9244890 DOI: 10.1038/s41541-022-00487-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.
Collapse
|
13
|
|
14
|
Kalnin KV, Plitnik T, Kishko M, Huang D, Raillard A, Piolat J, Anosova NG, Tibbitts T, DiNapoli J, Karve S, Goldman R, Gopani H, Dias A, Tran K, Zacharia M, Gu X, Boeglin L, Abysalh J, Vargas J, Beaulieu A, Shah M, Jeannotte T, Gillis K, Chivukula S, Swearingen R, Landolfi V, Fu TM, DeRosa F, Casimiro D. Pan-SARS neutralizing responses after third boost vaccination in non-human primate immunogenicity model. Vaccine 2022; 40:1289-1298. [PMID: 35101265 PMCID: PMC8801978 DOI: 10.1016/j.vaccine.2022.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/08/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022]
Abstract
The emergence of SARS-CoV-2 variants, especially Beta and Delta, has raised concerns about the reduced protection from previous infection or vaccination based on the original Wuhan-Hu-1 (D614) virus. To identify promising regimens for inducing neutralizing titers towards new variants, we evaluated monovalent and bivalent mRNA vaccines either as primary vaccination or as a booster in nonhuman primates (NHPs). Two mRNA vaccines, D614-based MRT5500 and Beta-based MRT5500β, tested in sequential regimens or as a bivalent combination in naïve NHPs produced modest neutralizing titers to heterologous variants. However, when mRNA vaccines were administered as a booster to pre-immune NHPs, we observed a robust increase in neutralizing titers with expanded breadth towards all tested variants, and notably SARS-CoV-1. The breadth of the neutralizing response was independent of vaccine sequence or modality, as we further showed either MRT5500 or recombinant subunit Spike protein (with adjuvant) can serve as boosters to induce broadly neutralizing antibodies in the NHPs primed with MRT5500. The data support the notion that a third vaccination is key to boosting existing titers and improving the breadth of antibodies to address variants of concern, including those with an E484K mutation in the Receptor Binding Domain (RBD) (Beta, Gamma).
Collapse
Affiliation(s)
- Kirill V Kalnin
- Emergent BioSolutions, 3985-A Sorrento Valley Blvd, San Diego, CA 92121, United States
| | - Timothy Plitnik
- Yoh Services LLC, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Michael Kishko
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Dean Huang
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Alice Raillard
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| | - Julie Piolat
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| | - Natalie G Anosova
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States.
| | - Timothy Tibbitts
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Joshua DiNapoli
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Shrirang Karve
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Rebecca Goldman
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Hardip Gopani
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Anusha Dias
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Khang Tran
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Minnie Zacharia
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Xiaobo Gu
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Lianne Boeglin
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Jonathan Abysalh
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Jorel Vargas
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Angela Beaulieu
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Monic Shah
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Travis Jeannotte
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Kimberly Gillis
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Sudha Chivukula
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, United States
| | - Ron Swearingen
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | | | - Tong-Ming Fu
- UT Health Science Center at Houston, 7000 Fannin St #1200, Houston, TX 77030, United States
| | - Frank DeRosa
- Translate Bio, 29 Hartwell Ave, Lexington, MA 02421, United States
| | - Danilo Casimiro
- Sanofi Pasteur, 1541 AV Marcel Mérieux, 69280 Marcy l'Etoile, France
| |
Collapse
|
15
|
Mendes A, Azevedo-Silva J, Fernandes JC. From Sharks to Yeasts: Squalene in the Development of Vaccine Adjuvants. Pharmaceuticals (Basel) 2022; 15:265. [PMID: 35337064 PMCID: PMC8951290 DOI: 10.3390/ph15030265] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Squalene is a natural linear triterpene that can be found in high amounts in certain fish liver oils, especially from deep-sea sharks, and to a lesser extent in a wide variety of vegeTable oils. It is currently used for numerous vaccine and drug delivery emulsions due to its stability-enhancing properties and biocompatibility. Squalene-based vaccine adjuvants, such as MF59 (Novartis), AS03 (GlaxoSmithKline Biologicals), or AF03 (Sanofi) are included in seasonal vaccines against influenza viruses and are presently being considered for inclusion in several vaccines against SARS-CoV-2 and future pandemic threats. However, harvesting sharks for this purpose raises serious ecological concerns that the exceptional demand of the pandemic has exacerbated. In this line, the use of plants to obtain phytosqualene has been seen as a more sustainable alternative, yet the lower yields and the need for huge investments in infrastructures and equipment makes this solution economically ineffective. More recently, the enormous advances in the field of synthetic biology provided innovative approaches to make squalene production more sustainable, flexible, and cheaper by using genetically modified microbes to produce pharmaceutical-grade squalene. Here, we review the biological mechanisms by which squalene-based vaccine adjuvants boost the immune response, and further compare the existing sources of squalene and their environmental impact. We propose that genetically engineered microbes are a sustainable alternative to produce squalene at industrial scale, which are likely to become the sole source of pharmaceutical-grade squalene in the foreseeable future.
Collapse
Affiliation(s)
- Adélia Mendes
- Centro de Biotecnologia e Química Fina (CBQF), Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Amyris Bio Products Portugal, 4169-005 Porto, Portugal; (J.A.-S.); (J.C.F.)
| | | | | |
Collapse
|
16
|
Yang JX, Tseng JC, Yu GY, Luo Y, Huang CYF, Hong YR, Chuang TH. Recent Advances in the Development of Toll-like Receptor Agonist-Based Vaccine Adjuvants for Infectious Diseases. Pharmaceutics 2022; 14:pharmaceutics14020423. [PMID: 35214155 PMCID: PMC8878135 DOI: 10.3390/pharmaceutics14020423] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Vaccines are powerful tools for controlling microbial infections and preventing epidemic diseases. Efficient inactive, subunit, or viral-like particle vaccines usually rely on a safe and potent adjuvant to boost the immune response to the antigen. After a slow start, over the last decade there has been increased developments on adjuvants for human vaccines. The development of adjuvants has paralleled our increased understanding of the molecular mechanisms for the pattern recognition receptor (PRR)-mediated activation of immune responses. Toll-like receptors (TLRs) are a group of PRRs that recognize microbial pathogens to initiate a host’s response to infection. Activation of TLRs triggers potent and immediate innate immune responses, which leads to subsequent adaptive immune responses. Therefore, these TLRs are ideal targets for the development of effective adjuvants. To date, TLR agonists such as monophosphoryl lipid A (MPL) and CpG-1018 have been formulated in licensed vaccines for their adjuvant activity, and other TLR agonists are being developed for this purpose. The COVID-19 pandemic has also accelerated clinical research of vaccines containing TLR agonist-based adjuvants. In this paper, we reviewed the agonists for TLR activation and the molecular mechanisms associated with the adjuvants’ effects on TLR activation, emphasizing recent advances in the development of TLR agonist-based vaccine adjuvants for infectious diseases.
Collapse
Affiliation(s)
- Jing-Xing Yang
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
| | - Jen-Chih Tseng
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan;
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
| | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Yi-Ren Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
- Department of Life Sciences, National Central University, Taoyuan City 32001, Taiwan
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-37-246166 (ext. 37611)
| |
Collapse
|
17
|
De Rosa SC, Cohen KW, Bonaparte M, Fu B, Garg S, Gerard C, Goepfert PA, Huang Y, Larocque D, McElrath MJ, Morris D, Van der Most R, de Bruyn G, Pagnon A. Whole-blood cytokine secretion assay as a high-throughput alternative for assessing the cell-mediated immunity profile after two doses of an adjuvanted SARS-CoV-2 recombinant protein vaccine candidate. Clin Transl Immunology 2022; 11:e1360. [PMID: 35035955 PMCID: PMC8752373 DOI: 10.1002/cti2.1360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/25/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES We previously described the Phase I-II evaluation of SARS-CoV-2 recombinant protein candidate vaccine, CoV2-PreS-dTM, with AF03- or AS03-adjuvant systems (ClinicalTrials.gov, NCT04537208). Here, we further characterise the cellular immunogenicity profile of this vaccine candidate using a whole-blood secretion assay in parallel to intracellular cytokine staining (ICS) of cryopreserved peripheral blood mononuclear cells (PBMCs). METHODS A randomly allocated subset of 90 healthy, SARS-CoV-2-seronegative adults aged ≥ 18 years who had received (random allocation) one or two separate injections (on study day [D]1 and D22) of saline placebo or CoV2-PreS-dTM formulated with AS03 or AF03 were included. Cytokine secretion was assessed using a TruCulture® whole-blood stimulation system in combination with multiplex bead array, and intracellular cytokine profiles were evaluated on thawed PBMCs following ex vivo stimulation with recombinant S protein at pre-vaccination (D1), post-dose 1 (D22) and post-dose 2 (D36). RESULTS Both methods detected similar vaccine-induced responses after the first and second doses. We observed a Th1 bias (Th1/Th2 ratio > 1.0) for most treatment groups when analysed in whole blood, mainly characterised by increased IFN-γ, IL-2 and TNF-α secretion. Among participants aged ≥ 50 years, the Th1/Th2 ratio was higher for those who received vaccine candidate with AS03 versus AF03 adjuvant. ICS revealed that this higher Th1/Th2 ratio resulted from higher levels of IFN-γ expression and that the vaccine induced polyfunctional CD4+ T cells. CONCLUSIONS The whole-blood cytokine secretion assay is a high-throughput alternative for assessing the quantity and character of vaccine-induced cellular responses.
Collapse
Affiliation(s)
- Stephen C De Rosa
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Kristen W Cohen
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - Bo Fu
- BiostaticsSanofi PasteurSwiftwaterPAUSA
| | | | | | - Paul A Goepfert
- Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Ying Huang
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - M. Juliana McElrath
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Daryl Morris
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - Guy de Bruyn
- Global Clinical DevelopmentSanofi PasteurSwiftwaterPAUSA
| | - Anke Pagnon
- Research DepartmentSanofi PasteurMarcy l’ÉtoileFrance
| |
Collapse
|
18
|
Khandker SS, Godman B, Jawad MI, Meghla BA, Tisha TA, Khondoker MU, Haq MA, Charan J, Talukder AA, Azmuda N, Sharmin S, Jamiruddin MR, Haque M, Adnan N. A Systematic Review on COVID-19 Vaccine Strategies, Their Effectiveness, and Issues. Vaccines (Basel) 2021; 9:1387. [PMID: 34960133 PMCID: PMC8708628 DOI: 10.3390/vaccines9121387] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 vaccines are indispensable, with the number of cases and mortality still rising, and currently no medicines are routinely available for reducing morbidity and mortality, apart from dexamethasone, although others are being trialed and launched. To date, only a limited number of vaccines have been given emergency use authorization by the US Food and Drug Administration and the European Medicines Agency. There is a need to systematically review the existing vaccine candidates and investigate their safety, efficacy, immunogenicity, unwanted events, and limitations. The review was undertaken by searching online databases, i.e., Google Scholar, PubMed, and ScienceDirect, with finally 59 studies selected. Our findings showed several types of vaccine candidates with different strategies against SARS-CoV-2, including inactivated, mRNA-based, recombinant, and nanoparticle-based vaccines, are being developed and launched. We have compared these vaccines in terms of their efficacy, side effects, and seroconversion based on data reported in the literature. We found mRNA vaccines appeared to have better efficacy, and inactivated ones had fewer side effects and similar seroconversion in all types of vaccines. Overall, global variant surveillance and systematic tweaking of vaccines, coupled with the evaluation and administering vaccines with the same or different technology in successive doses along with homologous and heterologous prime-booster strategy, have become essential to impede the pandemic. Their effectiveness appreciably outweighs any concerns with any adverse events.
Collapse
Affiliation(s)
- Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Brian Godman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G1 1XQ, UK;
- Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Md. Irfan Jawad
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Bushra Ayat Meghla
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Taslima Akter Tisha
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Mohib Ullah Khondoker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar 1344, Bangladesh
| | - Md. Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur 342005, India;
| | - Ali Azam Talukder
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Nafisa Azmuda
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Shahana Sharmin
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mohd. Raeed Jamiruddin
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sugai Besi, Kuala Lumpur 57000, Malaysia
| | - Nihad Adnan
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| |
Collapse
|
19
|
Lim JW, Ahn YR, Park G, Kim HO, Haam S. Application of Nanomaterials as an Advanced Strategy for the Diagnosis, Prevention, and Treatment of Viral Diseases. Pharmaceutics 2021; 13:1570. [PMID: 34683863 PMCID: PMC8540357 DOI: 10.3390/pharmaceutics13101570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/19/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
The coronavirus disease (COVID-19) pandemic poses serious global health concerns with the continued emergence of new variants. The periodic outbreak of novel emerging and re-emerging infectious pathogens has elevated concerns and challenges for the future. To develop mitigation strategies against infectious diseases, nano-based approaches are being increasingly applied in diagnostic systems, prophylactic vaccines, and therapeutics. This review presents the properties of various nanoplatforms and discusses their role in the development of sensors, vectors, delivery agents, intrinsic immunostimulants, and viral inhibitors. Advanced nanomedical applications for infectious diseases have been highlighted. Moreover, physicochemical properties that confer physiological advantages and contribute to the control and inhibition of infectious diseases have been discussed. Safety concerns limit the commercial production and clinical use of these technologies in humans; however, overcoming these limitations may enable the use of nanomaterials to resolve current infection control issues via application of nanomaterials as a platform for the diagnosis, prevention, and treatment of viral diseases.
Collapse
Affiliation(s)
- Jong-Woo Lim
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| | - Yu-Rim Ahn
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
- Biohealth-machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Geunseon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| | - Hyun-Ouk Kim
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
- Biohealth-machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| |
Collapse
|
20
|
Durán-Lobato M, López-Estévez AM, Cordeiro AS, Dacoba TG, Crecente-Campo J, Torres D, Alonso MJ. Nanotechnologies for the delivery of biologicals: Historical perspective and current landscape. Adv Drug Deliv Rev 2021; 176:113899. [PMID: 34314784 DOI: 10.1016/j.addr.2021.113899] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
Biological macromolecule-based therapeutics irrupted in the pharmaceutical scene generating a great hope due to their outstanding specificity and potency. However, given their susceptibility to degradation and limited capacity to overcome biological barriers new delivery technologies had to be developed for them to reach their targets. This review aims at analyzing the historical seminal advances that shaped the development of the protein/peptide delivery field, along with the emerging technologies on the lead of the current landscape. Particularly, focus is made on technologies with a potential for transmucosal systemic delivery of protein/peptide drugs, followed by approaches for the delivery of antigens as new vaccination strategies, and formulations of biological drugs in oncology, with special emphasis on mAbs. Finally, a discussion of the key challenges the field is facing, along with an overview of prospective advances are provided.
Collapse
|
21
|
Goepfert PA, Fu B, Chabanon AL, Bonaparte MI, Davis MG, Essink BJ, Frank I, Haney O, Janosczyk H, Keefer MC, Koutsoukos M, Kimmel MA, Masotti R, Savarino SJ, Schuerman L, Schwartz H, Sher LD, Smith J, Tavares-Da-Silva F, Gurunathan S, DiazGranados CA, de Bruyn G. Safety and immunogenicity of SARS-CoV-2 recombinant protein vaccine formulations in healthy adults: interim results of a randomised, placebo-controlled, phase 1-2, dose-ranging study. THE LANCET. INFECTIOUS DISEASES 2021; 21:1257-1270. [PMID: 33887209 PMCID: PMC8055206 DOI: 10.1016/s1473-3099(21)00147-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 01/02/2023]
Abstract
Background CoV2 preS dTM is a stabilised pre-fusion spike protein vaccine produced in a baculovirus expression system being developed against SARS-CoV-2. We present interim safety and immunogenicity results of the first-in-human study of the CoV2 preS dTM vaccine with two different adjuvant formulations. Methods This phase 1–2, randomised, double-blind study is being done in healthy, SARS-CoV-2-seronegative adults in ten clinical research centres in the USA. Participants were stratified by age (18–49 years and ≥50 years) and randomly assigned using an interactive response technology system with block randomisation (blocks of varying size) to receive one dose (on day 1) or two doses (on days 1 and 22) of placebo or candidate vaccine, containing low-dose (effective dose 1·3 μg) or high-dose (2·6 μg) antigen with adjuvant AF03 (Sanofi Pasteur) or AS03 (GlaxoSmithKline) or unadjuvanted high-dose antigen (18–49 years only). Primary endpoints were safety, assessed up to day 43, and immunogenicity, measured as SARS-C0V-2 neutralising antibodies (geometric mean titres), assessed on days 1, 22, and 36 serum samples. Safety was assessed according to treatment received in the safety analysis set, which included all randomly assigned participants who received at least one dose. Neutralising antibody titres were assessed in the per-protocol analysis set for immunogenicity, which included participants who received at least one dose, met all inclusion and exclusion criteria, had no protocol deviation, had negative results in the neutralisation test at baseline, and had at least one valid post-dose serology sample. This planned interim analysis reports data up to 43 days after the first vaccination; participants in the trial will be followed up for 12 months after the last study injection. This trial is registered with ClinicalTrials.gov, NCT04537208, and is ongoing. Findings Between Sept 3 and Sept 29, 2020, 441 individuals (299 aged 18–49 years and 142 aged ≥50 years) were randomly assigned to one of the 11 treatment groups. The interim safety analyses included 439 (>99%) of 441 randomly assigned participants (299 aged 18–49 years and 140 aged ≥50 years). Neutralising antibody titres were analysed in 326 (74%) of 441 participants (235 [79%] of 299 aged 18–49 years and 91 [64%] of 142 aged ≥50 years). There were no vaccine-related unsolicited immediate adverse events, serious adverse events, medically attended adverse events classified as severe, or adverse events of special interest. Among all study participants, solicited local and systemic reactions of any grade after two vaccine doses were reported in 81% (95% CI 61–93; 21 of 26) of participants in the low-dose plus AF03 group, 93% (84–97; 74 of 80) in the low-dose plus AS03 group, 89% (70–98; 23 of 26) in the high-dose plus AF03 group, 95% (88–99; 81 of 85) in the high-dose plus AS03 group, 29% (10–56; five of 17) in the unadjuvanted high-dose group, and 21% (8–40; six of 29) in the placebo group. A single vaccine dose did not generate neutralising antibody titres above placebo levels in any group at days 22 or 36. Among participants aged 18–49 years, neutralising antibody titres after two vaccine doses were 13·1 (95% CI 6·40–26·9) in the low-dose plus AF03 group, 20·5 (13·1–32·1) in the low-dose plus AS03 group, 43·2 (20·6–90·4) in the high-dose plus AF03 group, 75·1 (50·5–112·0) in the high-dose plus AS03 group, 5·00 (not calculated) in the unadjuvanted high-dose group, and 5·00 (not calculated) in the placebo group. Among participants aged 50 years or older, neutralising antibody titres after two vaccine doses were 8·62 (1·90–39·0) in the low-dose plus AF03 group, 12·9 (7·09–23·4) in the low-dose plus AS03 group, 12·3 (4·35–35·0) in the high-dose plus AF03 group, 52·3 (25·3–108·0) in the high-dose plus AS03 group, and 5·00 (not calculated) in the placebo group. Interpretation The lower than expected immune responses, especially in the older age groups, and the high reactogenicity after dose two were probably due to higher than anticipated host-cell protein content and lower than planned antigen doses in the formulations tested, which was discovered during characterisation studies on the final bulk drug substance. Further development of the AS03-adjuvanted candidate vaccine will focus on identifying the optimal antigen formulation and dose. Funding Sanofi Pasteur and Biomedical Advanced Research and Development Authority.
Collapse
Affiliation(s)
- Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, AL, USA
| | - Bo Fu
- Sanofi Pasteur, Swiftwater, PA, USA
| | | | | | | | | | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Michael C Keefer
- University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | | | | | | | | | | | | | - Lawrence D Sher
- Peninsula Research Associates, Rolling Hills Estates, CA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, Cohen JD, Singh PA, Baldi A, Bajwa N, Kumar R, Vora LK, Patel TA, Oleynikov MD, Soni D, Yeapuri P, Mukadam I, Chakraborty R, Saksena CG, Herskovitz J, Hasan M, Oupicky D, Das S, Donnelly RF, Hettie KS, Chang L, Gendelman HE, Kevadiya BD. Nanocarrier vaccines for SARS-CoV-2. Adv Drug Deliv Rev 2021; 171:215-239. [PMID: 33428995 PMCID: PMC7794055 DOI: 10.1016/j.addr.2021.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/18/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Srijanee Das
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Preet Amol Singh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Ashish Baldi
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Neha Bajwa
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Tapan A Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology (CHARUSAT), Changa, Anand 388421, Gujarat, India
| | - Maxim D Oleynikov
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Rajashree Chakraborty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Caroline G Saksena
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Suvarthi Das
- Department of Medicine, Stanford Medical School, Stanford University, Palo Alto, CA 94304, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University, Palo Alto, CA 94304, USA
| | - Linda Chang
- Departments of Diagnostic Radiology & Nuclear Medicine, and Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| |
Collapse
|
23
|
Nguyen-Contant P, Sangster MY, Topham DJ. Squalene-Based Influenza Vaccine Adjuvants and Their Impact on the Hemagglutinin-Specific B Cell Response. Pathogens 2021; 10:pathogens10030355. [PMID: 33802803 PMCID: PMC8002393 DOI: 10.3390/pathogens10030355] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 11/23/2022] Open
Abstract
Influenza infections continue to cause significant annual morbidity and mortality despite ongoing influenza vaccine research. Adjuvants are administered in conjunction with influenza vaccines to enhance the immune response and strengthen protection against disease. Squalene-based emulsion adjuvants including MF59, AS03, and AF03, are registered for administration with influenza vaccines and are widely used in many countries. Squalene-based emulsion adjuvants induce a strong innate immune response, enhancing antigen presentation both quantitively and qualitatively to generate strong B cell responses and antibody production. They also diversify the reactivity profiles and strengthen the affinities of antibodies against the influenza hemagglutinin, increasing protection across virus clades. In this review, we consider the mechanisms of the enhancement of innate and adaptive immune responses by squalene-based emulsionSE adjuvants and the resulting increase in magnitude and breadth of hemagglutinin-specific B cell responses. We relate observed effects of SE adjuvants and current mechanistic understandings to events in responding lymph nodes. These insights will guide the rational design and optimization of influenza vaccines to provide broad and effective protection.
Collapse
|
24
|
Darricarrère N, Qiu Y, Kanekiyo M, Creanga A, Gillespie RA, Moin SM, Saleh J, Sancho J, Chou TH, Zhou Y, Zhang R, Dai S, Moody A, Saunders KO, Crank MC, Mascola JR, Graham BS, Wei CJ, Nabel GJ. Broad neutralization of H1 and H3 viruses by adjuvanted influenza HA stem vaccines in nonhuman primates. Sci Transl Med 2021; 13:13/583/eabe5449. [PMID: 33658355 DOI: 10.1126/scitranslmed.abe5449] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
Seasonal influenza vaccines confer protection against specific viral strains but have restricted breadth that limits their protective efficacy. The H1 and H3 subtypes of influenza A virus cause most of the seasonal epidemics observed in humans and are the major drivers of influenza A virus-associated mortality. The consequences of pandemic spread of COVID-19 underscore the public health importance of prospective vaccine development. Here, we show that headless hemagglutinin (HA) stabilized-stem immunogens presented on ferritin nanoparticles elicit broadly neutralizing antibody (bnAb) responses to diverse H1 and H3 viruses in nonhuman primates (NHPs) when delivered with a squalene-based oil-in-water emulsion adjuvant, AF03. The neutralization potency and breadth of antibodies isolated from NHPs were comparable to human bnAbs and extended to mismatched heterosubtypic influenza viruses. Although NHPs lack the immunoglobulin germline VH1-69 residues associated with the most prevalent human stem-directed bnAbs, other gene families compensated to generate bnAbs. Isolation and structural analyses of vaccine-induced bnAbs revealed extensive interaction with the fusion peptide on the HA stem, which is essential for viral entry. Antibodies elicited by these headless HA stabilized-stem vaccines neutralized diverse H1 and H3 influenza viruses and shared a mode of recognition analogous to human bnAbs, suggesting that these vaccines have the potential to confer broadly protective immunity against diverse viruses responsible for seasonal and pandemic influenza infections in humans.
Collapse
Affiliation(s)
| | - Yu Qiu
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca A Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Syed M Moin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jose Sancho
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Te-Hui Chou
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Yanfeng Zhou
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Ruijun Zhang
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Shujia Dai
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA
| | - Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michelle C Crank
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chih-Jen Wei
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA.
| | - Gary J Nabel
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA.
| |
Collapse
|
25
|
Pavot V, Bisceglia H, Guillaume F, Montano S, Zhang L, Boudet F, Haensler J. A novel vaccine adjuvant based on straight polyacrylate potentiates vaccine-induced humoral and cellular immunity in cynomolgus macaques. Hum Vaccin Immunother 2021; 17:2336-2348. [PMID: 33427044 PMCID: PMC8189108 DOI: 10.1080/21645515.2020.1855956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Adjuvants are central to the efficacy of subunit vaccines. Although several new adjuvants have been approved in human vaccines over the last decade, the panel of adjuvants in licensed human vaccines remains small. There is still a need for novel adjuvants that can be safely used in humans, easy to source and to formulate with a wide range of antigens and would be broadly applicable to a wide range of vaccines. In this article, using the Respiratory Syncytial Virus (RSV) nanoparticulate prefusion F model antigen developed by Sanofi, we demonstrate in the macaque model that the polyacrylate (PAA)-based adjuvant SPA09 is well tolerated and increases vaccine antigen-specific humoral immunity (sustained neutralizing antibodies, memory B cells and mucosal immunity) and elicits strong TH1-type responses (based on IFNγ and IL-2 ELISpots) in a dose-dependent manner. These data warrant further development of the SPA09 adjuvant for evaluation in clinical trials.
Collapse
Affiliation(s)
- Vincent Pavot
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| | - Hélène Bisceglia
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| | - Florine Guillaume
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| | - Sandrine Montano
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| | - Linong Zhang
- Sanofi Pasteur, Research & Development Department, Cambridge, MA, USA
| | - Florence Boudet
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| | - Jean Haensler
- Sanofi Pasteur, Research & Development Department, Marcy L'Etoile, France
| |
Collapse
|
26
|
Swanson KA, Rainho-Tomko JN, Williams ZP, Lanza L, Peredelchuk M, Kishko M, Pavot V, Alamares-Sapuay J, Adhikarla H, Gupta S, Chivukula S, Gallichan S, Zhang L, Jackson N, Yoon H, Edwards D, Wei CJ, Nabel GJ. A respiratory syncytial virus (RSV) F protein nanoparticle vaccine focuses antibody responses to a conserved neutralization domain. Sci Immunol 2020; 5:5/47/eaba6466. [PMID: 32358170 DOI: 10.1126/sciimmunol.aba6466] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
A stabilized form of the respiratory syncytial virus (RSV) fusion (F) protein has been explored as a vaccine to prevent viral infection because it presents several potent neutralizing epitopes. Here, we used a structure-based rational design to optimize antigen presentation and focus antibody (Ab) responses to key epitopes on the pre-fusion (pre-F) protein. This protein was fused to ferritin nanoparticles (pre-F-NP) and modified with glycans to mask nonneutralizing or poorly neutralizing epitopes to further focus the Ab response. The multimeric pre-F-NP elicited durable pre-F-specific Abs in nonhuman primates (NHPs) after >150 days and elicited potent neutralizing Ab (NAb) responses in mice and NHPs in vivo, as well as in human cells evaluated in the in vitro MIMIC system. This optimized pre-F-NP stimulated a more potent Ab response than a representative pre-F trimer, DS-Cav1. Collectively, this pre-F vaccine increased the generation of NAbs targeting the desired pre-F conformation, an attribute that facilitates the development of an effective RSV vaccine.
Collapse
Affiliation(s)
| | | | - Zachary P Williams
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Lilibeth Lanza
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Michael Peredelchuk
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Michael Kishko
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Vincent Pavot
- Sanofi Pasteur, 1541 Avenue Marcel Mérieux, Marcy l'Etoile, France
| | | | | | - Sankalp Gupta
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | | | - Scott Gallichan
- Sanofi Pasteur, 95 Willowdale Blvd, Toronto, Ontario, Canada
| | - Linong Zhang
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | | | - Heesik Yoon
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Darin Edwards
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | | | - Gary J Nabel
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA.
| |
Collapse
|
27
|
Mukherjee S, Pradhan S, Ghosh S, Sundar S, Das S, Mukherjee B, Roy S. Short-Course Treatment With Imipramine Entrapped in Squalene Liposomes Results in Sterile Cure of Experimental Visceral Leishmaniasis Induced by Antimony Resistant Leishmania donovani With Increased Efficacy. Front Cell Infect Microbiol 2020; 10:595415. [PMID: 33240825 PMCID: PMC7683767 DOI: 10.3389/fcimb.2020.595415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/12/2020] [Indexed: 11/13/2022] Open
Abstract
Previously we have shown that long term oral treatment of tricyclic-antidepressant-drug, imipramine, against experimental visceral leishmaniasis, results in clearance of organ parasites, regardless of input infection, either with antimony-sensitive (SbS) or antimony-resistant (SbR) Leishmania donovani (LD) clinical isolates. Although continuous imipramine monotherapy for 28 days (5 mg/kg) results in significant clearance of organ parasites in both SbR and SbSLD infected hamsters, the dose for the sterile parasite clearance from visceral organ is comparatively higher (10 mg/kg) and shows signs of toxicity. Hence, to reduce the toxicity, we encapsulated imipramine in squalene-phosphatidylcholine (SP) liposome (Lip-Imi) and tested its efficacy for a short-course treatment (10 days) in the animal model of visceral leishmaniasis. We observed a significant reduction of hepatic toxicity coupled with sterile parasite clearance in case of this short-course treatment of Lip-Imi, which is absent with free Imi treatment. This also correlates with significant increase in serum availability of imipramine in case of Lip-Imi treatment due to sustained release. Clearance of parasite was coupled with the polarization of antileishmanial immune repertoire from Th2 to Th1 after treatment with Lip-Imi in both SbRLD and SbSLD infected mouse models of LD infection. This study showed that imipramine is effective against both SbSLD and SbRLD at a significantly lower dose with reduced time course of treatment without any toxic side effects, when encapsulated in SP-liposome. Thus, the drug has the potential to be repurposed for the treatment of Kala-azar.
Collapse
Affiliation(s)
- Sandip Mukherjee
- Infectious Disease and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Supratim Pradhan
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | - Souradeepa Ghosh
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shantanabha Das
- Infectious Disease and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Budhaditya Mukherjee
- Infectious Disease and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | - Syamal Roy
- Infectious Disease and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,National Institute of Pharmaceutical Education & Research, Kolkata, India
| |
Collapse
|
28
|
O'Hagan DT, Lodaya RN, Lofano G. The continued advance of vaccine adjuvants - 'we can work it out'. Semin Immunol 2020; 50:101426. [PMID: 33257234 DOI: 10.1016/j.smim.2020.101426] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
In the last decade there have been some significant advances in vaccine adjuvants, particularly in relation to their inclusion in licensed products. This was proceeded by several decades in which such advances were very scarce, or entirely absent, but several novel adjuvants have now been included in licensed products, including in the US. These advances have relied upon several key technological insights that have emerged in this time period, which have finally allowed an in depth understanding of how adjuvants work. These advances include developments in systems biology approaches which allow the hypotheses first advanced in pre-clinical studies to be critically evaluated in human studies. This review highlights these recent advances, both in relation to the adjuvants themselves, but also the technologies that have enabled their successes. Moreover, we critically appraise what will come next, both in terms of new adjuvant molecules, and the technologies needed to allow them to succeed. We confidently predict that additional adjuvants will emerge in the coming years that will reach approval in licensed products, but that the components might differ significantly from those which are currently used. Gradually, the natural products that were originally used to build adjuvants, since they were readily available at the time of initial development, will come to be replaced by synthetic or biosynthetic materials, with more appealing attributes, including more reliable and robust supply, along with reduced heterogeneity. The recent advance in vaccine adjuvants is timely, given the need to create novel vaccines to deal with the COVID-19 pandemic. Although, we must ensure that the rigorous safety evaluations that allowed the current adjuvants to advance are not 'short-changed' in the push for new vaccines to meet the global challenge as quickly as possible, we must not jeopardize what we have achieved, by pushing less established technologies too quickly, if the data does not fully support it.
Collapse
Affiliation(s)
- Derek T O'Hagan
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Rushit N Lodaya
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Giuseppe Lofano
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA.
| |
Collapse
|
29
|
Kheirollahpour M, Mehrabi M, Dounighi NM, Mohammadi M, Masoudi A. Nanoparticles and Vaccine Development. Pharm Nanotechnol 2020; 8:6-21. [PMID: 31647394 DOI: 10.2174/2211738507666191024162042] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/23/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022]
Abstract
In spite of the progress of conventional vaccines, improvements are required due to concerns about the low immunogenicity of the toxicity, instability, and the need for multiple administrations of the vaccines. To overcome the mentioned problems, nanotechnology has recently been incorporated into vaccine development. Nanotechnology increasingly plays an important role in vaccine development nanocarrier-based delivery systems that offer an opportunity to increase the cellular and humoral immune responses. The use of nanoparticles in vaccine formulations allows not only enhanced immunogenicity and stability of antigen, but also targeted delivery and slow release. Over the past decade, nanoscale size materials such as virus-like particles, liposomes, ISCOMs, polymeric, inorganic nanoparticles and emulsions have gained attention as potential delivery vehicles for vaccine antigens, which can both stabilize vaccine antigens and act as adjuvants. This advantage is attributable to the nanoscale particle size, which facilitates uptake by Antigen- Presenting Cells (APCs), then leading to efficient antigen recognition and presentation. Modifying the surfaces of nanoparticles with different targeting moieties permits the delivery of antigens to specific receptors on the cell surface, thereby stimulating selective and specific immune responses. This review provides an overview of recent advances in nanovaccinology.
Collapse
Affiliation(s)
- Mehdi Kheirollahpour
- Department of Human Vaccine and Serum, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.,Chemistry & Chemical Engineering Research Center of Iran, P.O. Box 14334-186, Tehran, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Naser Mohammadpour Dounighi
- Department of Human Vaccine and Serum, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mohsen Mohammadi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Masoudi
- Department of Pharmacology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
30
|
Evaluation of the respiratory syncytial virus G-directed neutralizing antibody response in the human airway epithelial cell model. Virology 2020; 550:21-26. [PMID: 32866728 DOI: 10.1016/j.virol.2020.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/08/2020] [Accepted: 08/17/2020] [Indexed: 11/21/2022]
Abstract
Human respiratory syncytial virus (RSV) is a major cause of serious respiratory tract infections in infants and the elderly. Recently it was shown that the RSV G glycoprotein mediates attachment to cells using CX3CR1 as a receptor, and that G-specific neutralizing antibodies can be detected using human airway epithelial (HAE) cell cultures. To investigate the contributions of G-specific antibodies to RSV neutralization, we performed HAE neutralization assays on sera from RSV G-immunized mice or RSV-infected infants. We confirmed that G-specific neutralization using serum from mice or humans could only be detected on HAE cultures. We also found that RSV G-specific antibodies in infants were either subgroup specific or cross-neutralizing. Altogether, our results suggest that G is an important target for generating neutralizing antibodies and would be beneficial to include in an RSV vaccine. Further, inclusion of G antigens from both RSV subgroups may enhance the vaccine cross protection potency.
Collapse
|
31
|
Weiss C, Carriere M, Fusco L, Capua I, Regla-Nava JA, Pasquali M, Scott JA, Vitale F, Unal MA, Mattevi C, Bedognetti D, Merkoçi A, Tasciotti E, Yilmazer A, Gogotsi Y, Stellacci F, Delogu LG. Toward Nanotechnology-Enabled Approaches against the COVID-19 Pandemic. ACS NANO 2020; 14:6383-6406. [PMID: 32519842 PMCID: PMC7299399 DOI: 10.1021/acsnano.0c03697] [Citation(s) in RCA: 342] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The COVID-19 outbreak has fueled a global demand for effective diagnosis and treatment as well as mitigation of the spread of infection, all through large-scale approaches such as specific alternative antiviral methods and classical disinfection protocols. Based on an abundance of engineered materials identifiable by their useful physicochemical properties through versatile chemical functionalization, nanotechnology offers a number of approaches to cope with this emergency. Here, through a multidisciplinary Perspective encompassing diverse fields such as virology, biology, medicine, engineering, chemistry, materials science, and computational science, we outline how nanotechnology-based strategies can support the fight against COVID-19, as well as infectious diseases in general, including future pandemics. Considering what we know so far about the life cycle of the virus, we envision key steps where nanotechnology could counter the disease. First, nanoparticles (NPs) can offer alternative methods to classical disinfection protocols used in healthcare settings, thanks to their intrinsic antipathogenic properties and/or their ability to inactivate viruses, bacteria, fungi, or yeasts either photothermally or via photocatalysis-induced reactive oxygen species (ROS) generation. Nanotechnology tools to inactivate SARS-CoV-2 in patients could also be explored. In this case, nanomaterials could be used to deliver drugs to the pulmonary system to inhibit interaction between angiotensin-converting enzyme 2 (ACE2) receptors and viral S protein. Moreover, the concept of "nanoimmunity by design" can help us to design materials for immune modulation, either stimulating or suppressing the immune response, which would find applications in the context of vaccine development for SARS-CoV-2 or in counteracting the cytokine storm, respectively. In addition to disease prevention and therapeutic potential, nanotechnology has important roles in diagnostics, with potential to support the development of simple, fast, and cost-effective nanotechnology-based assays to monitor the presence of SARS-CoV-2 and related biomarkers. In summary, nanotechnology is critical in counteracting COVID-19 and will be vital when preparing for future pandemics.
Collapse
Affiliation(s)
- Carsten Weiss
- Institute of Biological and Chemical
Systems, Biological Information Processing, Karlsruhe
Institute of Technology, Campus North,
Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen,
Germany
| | - Marie Carriere
- Univ. Grenoble
Alpes, CEA, CNRS, IRIG, SyMMES-CIBEST, F-38000
Grenoble, France
| | - Laura Fusco
- Department of Chemical and
Pharmaceutical Sciences, University of
Trieste, 34127 Trieste,
Italy
- Cancer Research Department,
Sidra Medicine, Doha,
Qatar
| | - Ilaria Capua
- One Health Center of Excellence,
University of Florida, Gainesville,
Florida 32611, United States
| | - Jose Angel Regla-Nava
- Division of Inflammation Biology,
La Jolla Institute for Allergy and
Immunology, La Jolla, California 92037,
United States
| | - Matteo Pasquali
- Department of Chemical &
Biomolecular Engineering, Rice University,
Houston, Texas 77251, United States
- Department of Chemistry,
Rice University, Houston, Texas
77251, United States
- Department of Materials Science and
Nanoengineering, Rice University, Houston,
Texas 77251, United States
| | - James A. Scott
- Dalla Lana School of Public Health,
University of Toronto, 223 College
Street, M5T 1R4 Toronto, Ontario, Canada
| | - Flavia Vitale
- Department of Neurology,
Bioengineering, Physical Medicine & Rehabilitation, Center for
Neuroengineering and Therapeutics, University of
Pennsylvania, Philadelphia, Pennsylvania 19104,
United States
- Center for Neurotrauma,
Neurodegeneration, and Restoration, Corporal Michael J.
Crescenz Veterans Affairs Medical Center,
Philadelphia, Pennsylvania 19104, United
States
| | | | - Cecilia Mattevi
- Department of Materials,
Imperial College London, London SW7
2AZ, United Kingdom
| | | | - Arben Merkoçi
- Nanobioelectronics & Biosensors
Group, Catalan Institute of Nanoscience and
Nanotechnology (ICN2), CSIC and BIST, Campus UAB,
08193 Bellaterra, Spain
- ICREA -
Institució Catalana de Recerca i Estudis
Avançats, ES-08010 Barcelona,
Spain
| | - Ennio Tasciotti
- Orthopedics and Sports Medicine,
Houston Methodist Hospital, Houston,
Texas 77030, United States
- Department of Plastic Surgery,
MD Anderson, Houston, Texas 77230,
United States
| | - Açelya Yilmazer
- Stem Cell Institute,
Ankara University, Ankara, 06100
Turkey
- Department of Biomedical Engineering,
Faculty of Engineering, Ankara University,
Ankara, 06100 Turkey
| | - Yury Gogotsi
- A.J. Drexel Nanomaterials Institute,
and Materials Science and Engineering Department, Drexel
University, Philadelphia, Pennsylvania 19104,
United States
| | - Francesco Stellacci
- Institute of Materials,
Ecole Polytechnique Federale de Lausanne
(EPFL), 1015 Lausanne,
Switzerland
- Interfaculty Bioengineering Institute,
Ecole Polytechnique Fédérale de
Lausanne (EPFL), 1015 Lausanne,
Switzerland
| | - Lucia Gemma Delogu
- Department of Biomedical Sciences,
University of Padua, 35122 Padova,
Italy
| |
Collapse
|
32
|
Saffarionpour S. Preparation of Food Flavor Nanoemulsions by High- and Low-Energy Emulsification Approaches. FOOD ENGINEERING REVIEWS 2019. [DOI: 10.1007/s12393-019-09201-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
33
|
Abstract
Nanotechnology, particularly nanoemulsions (NEs), have gained increasing interest from researchers throughout the years. The small-sized droplet with a high surface area makes NEs important in many industries. In this review article, the components, properties, formation, and applications are summarized. The advantages and disadvantages are also described in this article. The formation of the nanosized emulsion can be divided into two types: high and low energy methods. In high energy methods, high-pressure homogenization, microfluidization, and ultrasonic emulsification are described thoroughly. Spontaneous emulsification, phase inversion temperature (PIT), phase inversion composition (PIC), and the less known D-phase emulsification (DPE) methods are emphasized in low energy methods. The applications of NEs are described in three main areas which are food, cosmetics, and drug delivery.
Collapse
|
34
|
Rudicell RS, Garinot M, Kanekiyo M, Kamp HD, Swanson K, Chou TH, Dai S, Bedel O, Simard D, Gillespie RA, Yang K, Reardon M, Avila LZ, Besev M, Dhal PK, Dharanipragada R, Zheng L, Duan X, Dinapoli J, Vogel TU, Kleanthous H, Mascola JR, Graham BS, Haensler J, Wei CJ, Nabel GJ. Comparison of adjuvants to optimize influenza neutralizing antibody responses. Vaccine 2019; 37:6208-6220. [PMID: 31493950 DOI: 10.1016/j.vaccine.2019.08.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/26/2019] [Accepted: 08/17/2019] [Indexed: 12/14/2022]
Abstract
Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.
Collapse
Affiliation(s)
| | | | - Masaru Kanekiyo
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Rebecca A Gillespie
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
35
|
Ciabattini A, Nardini C, Santoro F, Garagnani P, Franceschi C, Medaglini D. Vaccination in the elderly: The challenge of immune changes with aging. Semin Immunol 2019; 40:83-94. [PMID: 30501873 DOI: 10.1016/j.smim.2018.10.010] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
Abstract
The unprecedented increase of life expectancy challenges society to protect the elderly from morbidity and mortality making vaccination a crucial mean to safeguard this population. Indeed, infectious diseases, such as influenza and pneumonia, are among the top killers of elderly people in the world. Elderly individuals are more prone to severe infections and less responsive to vaccination prevention, due to immunosenescence combined with the progressive increase of a proinflammatory status characteristic of the aging process (inflammaging). These factors are responsible for most age-related diseases and correlate with poor response to vaccination. Therefore, it is of utmost interest to deepen the knowledge regarding the role of inflammaging in vaccination responsiveness to support the development of effective vaccination strategies designed for elderly. In this review we analyse the impact of age-associated factors such as inflammaging, immunosenescence and immunobiography on immune response to vaccination in the elderly, and we consider systems biology approaches as a mean for integrating a multitude of data in order to rationally design vaccination approaches specifically tailored for the elderly.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Christine Nardini
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, SE-171 77, Stockholm, Sweden; Personal Genomics S.r.l., Via Roveggia, 43B, 37134, Verona, Italy; CNR IAC "Mauro Picone", Via dei Taurini, 19, 00185, Roma, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Paolo Garagnani
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, SE-171 77, Stockholm, Sweden; Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Via G. Petroni 26, 40139, Bologna, Italy; Department of Experimental, Diagnostic and Specialty Medicine (DIMES) - University of Bologna,40139, Bologna, Italy
| | - Claudio Franceschi
- IRCCS, Institute of Neurological Sciences of Bologna, Via Altura 3, 40139, Bologna, Italy.
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy.
| |
Collapse
|
36
|
Tayeb HH, Sainsbury F. Nanoemulsions in drug delivery: formulation to medical application. Nanomedicine (Lond) 2018; 13:2507-2525. [DOI: 10.2217/nnm-2018-0088] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Nanoscale oil-in-water emulsions (NEs), heterogeneous systems of two immiscible liquids stabilized by emulsifiers or surfactants, show great potential in medical applications because of their attractive characteristics for drug delivery. NEs have been explored as therapeutic carriers for hydrophobic compounds via various routes of administration. NEs provide opportunities to improve drug delivery via alternative administration routes. However, deep understanding of the NE manufacturing and functionalization fundamentals, and how they relate to the choice of administration route and pharmacological profile is still needed to ease the clinical translation of NEs. Here, we review the diversity of medical applications for NEs and how that governs their formulation, route of administration, and the emergence of increasing sophistication in NE design for specific application.
Collapse
Affiliation(s)
- Hossam H Tayeb
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Faculty of Applied Medical Sciences, King Abdul Abdul-Aziz University, Jeddah, Kingdom of Saudi Arabia
| | - Frank Sainsbury
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
37
|
Abstract
In spite of current influenza vaccines being immunogenic, evolution of the influenza virus can reduce efficacy and so influenza remains a major threat to public health. One approach to improve influenza vaccines is to include adjuvants; substances that boost the immune response. Adjuvants are particularly beneficial for influenza vaccines administered during a pandemic when a rapid response is required or for use in patients with impaired immune responses, such as infants and the elderly. This review outlines the current use of adjuvants in human influenza vaccines, including what they are, why they are used and what is known of their mechanism of action. To date, six adjuvants have been used in licensed human vaccines: Alum, MF59, AS03, AF03, virosomes and heat labile enterotoxin (LT). In general these adjuvants are safe and well tolerated, but there have been some rare adverse events when adjuvanted vaccines are used at a population level that may discourage the inclusion of adjuvants in influenza vaccines, for example the association of LT with Bell's Palsy. Improved understanding about the mechanisms of the immune response to vaccination and infection has led to advances in adjuvant technology and we describe the experimental adjuvants that have been tested in clinical trials for influenza but have not yet progressed to licensure. Adjuvants alone are not sufficient to improve influenza vaccine efficacy because they do not address the underlying problem of mismatches between circulating virus and the vaccine. However, they may contribute to improved efficacy of next-generation influenza vaccines and will most likely play a role in the development of effective universal influenza vaccines, though what that role will be remains to be seen.
Collapse
Affiliation(s)
- John S Tregoning
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ryan F Russell
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ekaterina Kinnear
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| |
Collapse
|
38
|
Fabrication of Transparent Lemon Oil Loaded Microemulsions by Phase Inversion Temperature (PIT) Method: Effect of Oil Phase Composition and Stability after Dilution. FOOD BIOPHYS 2017. [DOI: 10.1007/s11483-017-9480-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
39
|
Howard LM, Hoek KL, Goll JB, Samir P, Galassie A, Allos TM, Niu X, Gordy LE, Creech CB, Prasad N, Jensen TL, Hill H, Levy SE, Joyce S, Link AJ, Edwards KM. Cell-Based Systems Biology Analysis of Human AS03-Adjuvanted H5N1 Avian Influenza Vaccine Responses: A Phase I Randomized Controlled Trial. PLoS One 2017; 12:e0167488. [PMID: 28099485 PMCID: PMC5242433 DOI: 10.1371/journal.pone.0167488] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/15/2016] [Indexed: 12/28/2022] Open
Abstract
Background Vaccine development for influenza A/H5N1 is an important public health priority, but H5N1 vaccines are less immunogenic than seasonal influenza vaccines. Adjuvant System 03 (AS03) markedly enhances immune responses to H5N1 vaccine antigens, but the underlying molecular mechanisms are incompletely understood. Objective and Methods We compared the safety (primary endpoint), immunogenicity (secondary), gene expression (tertiary) and cytokine responses (exploratory) between AS03-adjuvanted and unadjuvanted inactivated split-virus H5N1 influenza vaccines. In a double-blinded clinical trial, we randomized twenty adults aged 18–49 to receive two doses of either AS03-adjuvanted (n = 10) or unadjuvanted (n = 10) H5N1 vaccine 28 days apart. We used a systems biology approach to characterize and correlate changes in serum cytokines, antibody titers, and gene expression levels in six immune cell types at 1, 3, 7, and 28 days after the first vaccination. Results Both vaccines were well-tolerated. Nine of 10 subjects in the adjuvanted group and 0/10 in the unadjuvanted group exhibited seroprotection (hemagglutination inhibition antibody titer > 1:40) at day 56. Within 24 hours of AS03-adjuvanted vaccination, increased serum levels of IL-6 and IP-10 were noted. Interferon signaling and antigen processing and presentation-related gene responses were induced in dendritic cells, monocytes, and neutrophils. Upregulation of MHC class II antigen presentation-related genes was seen in neutrophils. Three days after AS03-adjuvanted vaccine, upregulation of genes involved in cell cycle and division was detected in NK cells and correlated with serum levels of IP-10. Early upregulation of interferon signaling-related genes was also found to predict seroprotection 56 days after first vaccination. Conclusions Using this cell-based systems approach, novel mechanisms of action for AS03-adjuvanted pandemic influenza vaccination were observed. Trial Registration ClinicalTrials.gov NCT01573312
Collapse
Affiliation(s)
- Leigh M. Howard
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Kristen L. Hoek
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | | | - Parimal Samir
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Allison Galassie
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America
| | - Tara M. Allos
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Xinnan Niu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Laura E. Gordy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - C. Buddy Creech
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology; Huntsville, AL, United States of America
| | | | - Heather Hill
- The Emmes Corporation, Rockville, MD, United States of America
| | - Shawn E. Levy
- HudsonAlpha Institute for Biotechnology; Huntsville, AL, United States of America
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, United States of America
| | - Andrew J. Link
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- * E-mail: (KME); (AJL)
| | - Kathryn M. Edwards
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- * E-mail: (KME); (AJL)
| |
Collapse
|
40
|
Abstract
Emulsion adjuvants for human vaccines have evolved gradually over the last century. Current formulations are the result of many refinements to their composition and manufacturing, as well as optimization for safety and efficacy. Squalene has emerged as being particularly suitable for the manufacturing of safe oil-in-water (O/W) adjuvants for parenteral applications due to its biocompatibility and ability to be metabolized. Emulsion particle size has been identified as an important parameter affecting the pharmaceutical performance of O/W emulsion adjuvants. Submicronic emulsions with sizes in the 80-200 nm range are preferred for potency, manufacturing consistency, and stability reasons. Two manufacturing processes, high pressure homogenization (HPH or microfluidization) and a phase inversion temperature method (PIT), are described to yield such fine and long-term stable emulsion adjuvants.
Collapse
|
41
|
Abstract
Many human vaccines contain certain insoluble aluminum salts such as aluminum oxyhydroxide and aluminum hydroxyphosphate as vaccine adjuvants to boost the immunogenicity of the vaccines. Aluminum salts have been used as vaccine adjuvants for decades and have an established, favorable safety profile. However, preparing aluminum salts and aluminum salt-adjuvanted vaccines in a consistent manner remains challenging. This chapter discusses methods to prepare aluminum salts and aluminum salt-adjuvanted vaccines, factors to consider during preparation, and methods to characterize the vaccines after preparation.
Collapse
|
42
|
Calligaris S, Valoppi F, Barba L, Pizzale L, Anese M, Conte L, Nicoli MC. Development of Transparent Curcumin Loaded Microemulsions by Phase Inversion Temperature (PIT) Method: Effect of Lipid Type and Physical State on Curcumin Stability. FOOD BIOPHYS 2016. [DOI: 10.1007/s11483-016-9461-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Gaudin A, Song E, King AR, Saucier-Sawyer JK, Bindra R, Desmaële D, Couvreur P, Saltzman WM. PEGylated squalenoyl-gemcitabine nanoparticles for the treatment of glioblastoma. Biomaterials 2016; 105:136-144. [PMID: 27521616 DOI: 10.1016/j.biomaterials.2016.07.037] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 11/25/2022]
Abstract
New treatments for glioblastoma multiforme (GBM) are desperately needed, as GBM prognosis remains poor, mainly due to treatment resistance, poor distribution of therapeutics in the tumor tissue, and fast metabolism of chemotherapeutic drugs in the brain extracellular space. Convection-enhanced delivery (CED) of nanoparticles (NPs) has been shown to improve the delivery of chemotherapeutic drugs to the tumor bed, providing sustained release, and enhancing survival of animals with intracranial tumors. Here we administered gemcitabine, a nucleoside analog used as a first line treatment for a wide variety of extracranial solid tumors, within squalene-based NPs using CED, to overcome the above-mentioned challenges of GBM treatment. Small percentages of poly(ethylene) glycol (PEG) dramatically enhanced the distribution of squalene-gemcitabine nanoparticles (SQ-Gem NPs) in healthy animals and tumor-bearing animals after administration by CED. When tested in an orthotopic model of GBM, SQ-Gem-PEG NPs demonstrated significantly improved therapeutic efficacy compared to free gemcitabine, both as a chemotherapeutic drug and as a radiosensitizer. Furthermore, MR contrast agents were incorporated into the SQ-Gem-PEG NP formulation, providing a way to non-invasively track the NPs during infusion.
Collapse
Affiliation(s)
- Alice Gaudin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Eric Song
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Amanda R King
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | | | - Ranjit Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Didier Desmaële
- Institut Galien Paris-Sud, UMR CNRS 8612, University Paris-Sud XI, Châtenay-Malabry, 92290, France
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, University Paris-Sud XI, Châtenay-Malabry, 92290, France
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
44
|
Iyer V, Cayatte C, Guzman B, Schneider-Ohrum K, Matuszak R, Snell A, Rajani GM, McCarthy MP, Muralidhara B. Impact of formulation and particle size on stability and immunogenicity of oil-in-water emulsion adjuvants. Hum Vaccin Immunother 2016; 11:1853-64. [PMID: 26090563 DOI: 10.1080/21645515.2015.1046660] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Oil-in-water emulsions have gained consideration as vaccine adjuvants in recent years due to their ability to elicit a differentiated immunogenic response compared to traditional aluminum salt adjuvants. Squalene, a cholesterol precursor, is a natural product with immunostimulatory properties, making it an ideal candidate for such oil-in-water emulsions. Particle size is a key parameter of these emulsions and its relationship to stability and adjuvanticity has not been extensively studied. This study evaluates the effect of particle size on the stability and immunogenicity of squalene emulsions. We investigated the effect of formulation parameters such as surfactant concentration on particle size, resulting in particles with average diameter of 80 nm, 100 nm, 150 nm, 200 nm, or 250 nm. Emulsions were exposed to shear and temperature stresses, and stability parameters such as pH, osmolarity, size, and in-depth visual appearance were monitored over time. In addition, adjuvanticity of different particle size was assessed in a mouse model using Respiratory Syncytial Virus Fusion protein (RSV-F) as a model antigen. Temperature dependent phase separation appeared to be the most common route of degradation occurring in the higher particle sizes emulsions. The emulsions below 150 nm size maintained stability at either 5 °C or 25 °C, and the 80 nm diameter ones showed no measurable changes in size even after one month at 40 °C. In vivo studies using the emulsions as an adjuvant with RSV F antigen revealed that superior immunogenicity could be achieved with the 80 nm particle size emulsion.
Collapse
Affiliation(s)
- Vidyashankara Iyer
- a Formulation Sciences; Biopharmaceutical Development; Medimmune LLC ; Gaithersburg , MD USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Heo W, Kim JH, Pan JH, Kim YJ. Lecithin-Based Nano-emulsification Improves the Bioavailability of Conjugated Linoleic Acid. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:1355-1360. [PMID: 26822835 DOI: 10.1021/acs.jafc.5b05397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this study, we investigated the effects of lecithin-based nano-emulsification on the heat stability and bioavailability of conjugated linoleic acid (CLA) in different free fatty acid (FFA) and triglyceride (TG) forms. CLA nano-emulsion in TG form exhibited a small droplet size (70-120 nm) compared to CLA nano-emulsion in FFA form (230-260 nm). Nano-emulsification protected CLA isomers in TG form, but not in free form, against thermal decomposition during the heat treatment. The in vitro bioavailability test using monolayers of Caco-2 human intestinal cells showed that nano-emulsification increased the cellular uptake of CLA in both FFA and TG forms. More importantly, a rat feeding study showed that CLA content in small intestinal tissues or plasma was higher when CLA was emulsified, indicating an enhanced oral bioavailability of CLA by nano-emulsification. These results provide important information for development of nano-emulsion-based delivery systems that improve thermal stability and bioavailability of CLA.
Collapse
Affiliation(s)
- Wan Heo
- Department of Food and Biotechnology, Korea University , Sejong 30019, Republic of Korea
| | - Jun Ho Kim
- Department of Food and Biotechnology, Korea University , Sejong 30019, Republic of Korea
| | - Jeong Hoon Pan
- Department of Food and Biotechnology, Korea University , Sejong 30019, Republic of Korea
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University , Sejong 30019, Republic of Korea
| |
Collapse
|
46
|
On vaccine's adjuvants and autoimmunity: Current evidence and future perspectives. Autoimmun Rev 2015; 14:880-8. [DOI: 10.1016/j.autrev.2015.05.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/24/2015] [Indexed: 01/08/2023]
|
47
|
Haensler J, Probeck P, Su J, Piras F, Dalençon F, Cotte JF, Chambon V, Iqbal SM, Hawkins L, Burdin N. Design and preclinical characterization of a novel vaccine adjuvant formulation consisting of a synthetic TLR4 agonist in a thermoreversible squalene emulsion. Int J Pharm 2015; 486:99-111. [PMID: 25794609 DOI: 10.1016/j.ijpharm.2015.03.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/22/2022]
Abstract
We describe the development, analytical characterization, stability and preclinical efficacy of AF04, a combination adjuvant comprising the synthetic toll-like receptor 4 (TLR4) agonist, E6020, formulated in AF03, a thermoreversible squalene emulsion. By using AF04 with the recombinant major outer membrane protein of Chlamydia trachomatis (Ct-MOMP) and with the recombinant surface glycoprotein gB from human cytomegalovirus (CMV-gB) as model antigens, we show that AF03 and E6020 can synergize to augment specific antibody and Th-1 cellular immune responses in mice. In terms of formulation, we observe that the method used to incorporate E6020 into AF03 affects its partition between the oil and water phases of the emulsion which in turn has a significant impact on the tolerability (IV pyrogenicity test in rabbits) of this novel adjuvant combination.
Collapse
Affiliation(s)
| | | | - Jin Su
- Sanofi Pasteur R&D, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim MG, Park JY, Shon Y, Kim G, Shim G, Oh YK. Nanotechnology and vaccine development. Asian J Pharm Sci 2014. [DOI: 10.1016/j.ajps.2014.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
49
|
Garai P, Gogoi M, Gopal G, Radhakrishnan Y, Nandakumar KS, Chakravortty D. The basics and advances of immunomodulators and antigen presentation: a key to development of potent memory response against pathogens. Expert Opin Biol Ther 2014; 14:1383-97. [PMID: 24897303 DOI: 10.1517/14712598.2014.925871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Immunomodulators are agents, which can modulate the immune response to specific antigens, while causing least toxicity to the host system. Being part of the modern vaccine formulations, these compounds have contributed remarkably to the field of therapeutics. Despite the successful record maintained by these agents, the requirement of novel immunomodulators keeps increasing due to the increasing severity of diseases. Hence, research regarding the same holds great importance. AREAS COVERED In this review, we discuss the role of immunomodulators in improving performance of various vaccines used for counteracting most threatening infectious diseases, mechanisms behind their action and criteria for development of novel immunomodulators. EXPERT OPINION Understanding the molecular mechanisms underlying immune response is a prerequisite for development of effective therapeutics as these are often exploited by pathogens for their own propagation. Keeping this in mind, the present research in the field of immunotherapy focuses on developing immunomodulators that would not only enhance the protection against pathogen, but also generate a long-term memory response. With the introduction of advanced formulations including combination of different kinds of immunomodulators, one can expect tremendous success in near future.
Collapse
Affiliation(s)
- Preeti Garai
- Indian Institute of Science, Department of Microbiology and Cell Biology , Bangalore, 560012 , India +91 80 2293 2842 ; +91 80 2360 2697 ;
| | | | | | | | | | | |
Collapse
|
50
|
Fox CB, Haensler J. An update on safety and immunogenicity of vaccines containing emulsion-based adjuvants. Expert Rev Vaccines 2014; 12:747-58. [PMID: 23885820 DOI: 10.1586/14760584.2013.811188] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the exception of alum, emulsion-based vaccine adjuvants have been administered to far more people than any other adjuvant, especially since the 2009 H1N1 influenza pandemic. The number of clinical safety and immunogenicity evaluations of vaccines containing emulsion adjuvants has correspondingly mushroomed. In this review, the authors introduce emulsion adjuvant composition and history before detailing the most recent findings from clinical and postmarketing data regarding the effects of emulsion adjuvants on vaccine immunogenicity and safety, with emphasis on the most widely distributed emulsion adjuvants, MF59® and AS03. The authors also present a summary of other emulsion adjuvants in clinical development and indicate promising avenues for future emulsion-based adjuvant development. Overall, emulsion adjuvants have demonstrated potent adjuvant activity across a number of disease indications along with acceptable safety profiles.
Collapse
|